Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 138(5): 976-89, 2009 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-19737523

RESUMEN

Leptin inhibition of bone mass accrual requires the integrity of specific hypothalamic neurons but not expression of its receptor on these neurons. The same is true for its regulation of appetite and energy expenditure. This suggests that leptin acts elsewhere in the brain to achieve these three functions. We show here that brainstem-derived serotonin (BDS) favors bone mass accrual following its binding to Htr2c receptors on ventromedial hypothalamic neurons and appetite via Htr1a and 2b receptors on arcuate neurons. Leptin inhibits these functions and increases energy expenditure because it reduces serotonin synthesis and firing of serotonergic neurons. Accordingly, while abrogating BDS synthesis corrects the bone, appetite and energy expenditure phenotypes caused by leptin deficiency, inactivation of the leptin receptor in serotonergic neurons recapitulates them fully. This study modifies the map of leptin signaling in the brain and identifies a molecular basis for the common regulation of bone and energy metabolisms. For a video summary of this article, see the PaperFlick file with the Supplemental Data available online.


Asunto(s)
Apetito , Densidad Ósea , Metabolismo Energético , Leptina/metabolismo , Serotonina/metabolismo , Tronco Encefálico/metabolismo , Hipotálamo/metabolismo , Receptores de Leptina/metabolismo , Transducción de Señal
2.
Cell ; 135(5): 825-37, 2008 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-19041748

RESUMEN

Loss- and gain-of-function mutations in the broadly expressed gene Lrp5 affect bone formation, causing osteoporosis and high bone mass, respectively. Although Lrp5 is viewed as a Wnt coreceptor, osteoblast-specific disruption of beta-Catenin does not affect bone formation. Instead, we show here that Lrp5 inhibits expression of Tph1, the rate-limiting biosynthetic enzyme for serotonin in enterochromaffin cells of the duodenum. Accordingly, decreasing serotonin blood levels normalizes bone formation and bone mass in Lrp5-deficient mice, and gut- but not osteoblast-specific Lrp5 inactivation decreases bone formation in a beta-Catenin-independent manner. Moreover, gut-specific activation of Lrp5, or inactivation of Tph1, increases bone mass and prevents ovariectomy-induced bone loss. Serotonin acts on osteoblasts through the Htr1b receptor and CREB to inhibit their proliferation. By identifying duodenum-derived serotonin as a hormone inhibiting bone formation in an Lrp5-dependent manner, this study broadens our understanding of bone remodeling and suggests potential therapies to increase bone mass.


Asunto(s)
Duodeno/metabolismo , Proteínas Relacionadas con Receptor de LDL/metabolismo , Osteogénesis , Serotonina/metabolismo , Animales , Proteína de Unión a CREB/metabolismo , Femenino , Proteínas Relacionadas con Receptor de LDL/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones , Receptor de Serotonina 5-HT1B/metabolismo , Triptófano Hidroxilasa/metabolismo
3.
Proc Natl Acad Sci U S A ; 116(22): 11028-11037, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-31072928

RESUMEN

Mitochondria in neurons, in addition to their primary role in bioenergetics, also contribute to specialized functions, including regulation of synaptic transmission, Ca2+ homeostasis, neuronal excitability, and stress adaptation. However, the factors that influence mitochondrial biogenesis and function in neurons remain poorly elucidated. Here, we identify an important role for serotonin (5-HT) as a regulator of mitochondrial biogenesis and function in rodent cortical neurons, via a 5-HT2A receptor-mediated recruitment of the SIRT1-PGC-1α axis, which is relevant to the neuroprotective action of 5-HT. We found that 5-HT increased mitochondrial biogenesis, reflected through enhanced mtDNA levels, mitotracker staining, and expression of mitochondrial components. This resulted in higher mitochondrial respiratory capacity, oxidative phosphorylation (OXPHOS) efficiency, and a consequential increase in cellular ATP levels. Mechanistically, the effects of 5-HT were mediated via the 5-HT2A receptor and master modulators of mitochondrial biogenesis, SIRT1 and PGC-1α. SIRT1 was required to mediate the effects of 5-HT on mitochondrial biogenesis and function in cortical neurons. In vivo studies revealed that 5-HT2A receptor stimulation increased cortical mtDNA and ATP levels in a SIRT1-dependent manner. Direct infusion of 5-HT into the neocortex and chemogenetic activation of 5-HT neurons also resulted in enhanced mitochondrial biogenesis and function in vivo. In cortical neurons, 5-HT enhanced expression of antioxidant enzymes, decreased cellular reactive oxygen species, and exhibited neuroprotection against excitotoxic and oxidative stress, an effect that required SIRT1. These findings identify 5-HT as an upstream regulator of mitochondrial biogenesis and function in cortical neurons and implicate the mitochondrial effects of 5-HT in its neuroprotective action.


Asunto(s)
Mitocondrias , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Receptor de Serotonina 5-HT2A , Serotonina , Sirtuina 1 , Animales , Corteza Cerebral/citología , Masculino , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neuronas/citología , Neuronas/fisiología , Biogénesis de Organelos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT2A/genética , Receptor de Serotonina 5-HT2A/metabolismo , Serotonina/metabolismo , Serotonina/farmacología , Sirtuina 1/genética , Sirtuina 1/metabolismo
4.
Neuroimage ; 239: 118284, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34147630

RESUMEN

Resting functional MRI studies of the infant brain are increasingly becoming an important tool in developmental neuroscience. Whereas the test-retest reliability of functional connectivity (FC) measures derived from resting fMRI data have been characterized in the adult and child brain, similar assessments have not been conducted in infants. In this study, we examined the intra-session test-retest reliability of FC measures from 119 infant brain MRI scans from four neurodevelopmental studies. We investigated edge-level and subject-level reliability within one MRI session (between and within runs) measured by the Intraclass correlation coefficient (ICC). First, using an atlas-based approach, we examined whole-brain connectivity as well as connectivity within two common resting fMRI networks - the default mode network (DMN) and the sensorimotor network (SMN). Second, we examined the influence of run duration, study site, and scanning manufacturer (e.g., Philips and General Electric) on ICCs. Lastly, we tested spatial similarity using the Jaccard Index from networks derived from independent component analysis (ICA). Consistent with resting fMRI studies from adults, our findings indicated poor edge-level reliability (ICC = 0.14-0.18), but moderate-to-good subject-level intra-session reliability for whole-brain, DMN, and SMN connectivity (ICC = 0.40-0.78). We also found significant effects of run duration, site, and scanning manufacturer on reliability estimates. Some ICA-derived networks showed strong spatial reproducibility (e.g., DMN, SMN, and Visual Network), and were labelled based on their spatial similarity to analogous networks measured in adults. These networks were reproducibly found across different study sites. However, other ICA-networks (e.g. Executive Control Network) did not show strong spatial reproducibility, suggesting that the reliability and/or maturational course of functional connectivity may vary by network. In sum, our findings suggest that developmental scientists may be on safe ground examining the functional organization of some major neural networks (e.g. DMN and SMN), but judicious interpretation of functional connectivity is essential to its ongoing success.


Asunto(s)
Conectoma , Lactante , Imagen por Resonancia Magnética/métodos , Red Nerviosa/fisiología , Análisis por Conglomerados , Conjuntos de Datos como Asunto , Red en Modo Predeterminado , Femenino , Humanos , Masculino , Reproducibilidad de los Resultados , Descanso/fisiología
5.
Mol Psychiatry ; 25(12): 3304-3321, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-30120415

RESUMEN

Serotonin (5-HT) selective reuptake inhibitors (SSRIs) are widely used in the treatment of depression and anxiety disorders, but responsiveness is uncertain and side effects often lead to discontinuation. Side effect profiles suggest that SSRIs reduce dopaminergic (DAergic) activity, but specific mechanistic insight is missing. Here we show in mice that SSRIs impair motor function by acting on 5-HT2C receptors in the substantia nigra pars reticulata (SNr), which in turn inhibits nigra pars compacta (SNc) DAergic neurons. SSRI-induced motor deficits can be reversed by systemic or SNr-localized 5-HT2C receptor antagonism. SSRIs induce SNr hyperactivity and SNc hypoactivity that can also be reversed by systemic 5-HT2C receptor antagonism. Optogenetic inhibition of SNc DAergic neurons mimics the motor deficits due to chronic SSRI treatment, whereas local SNr 5-HT2C receptor antagonism or optogenetic activation of SNc DAergic neurons reverse SSRI-induced motor deficits. Lastly, we find that 5-HT2C receptor antagonism potentiates the antidepressant and anxiolytic effects of SSRIs. Together our findings demonstrate opposing roles for 5-HT2C receptors in the effects of SSRIs on motor function and affective behavior, highlighting the potential benefits of 5-HT2C receptor antagonists for both reduction of motor side effects of SSRIs and augmentation of therapeutic antidepressant and anxiolytic effects.


Asunto(s)
Receptor de Serotonina 5-HT2C , Inhibidores Selectivos de la Recaptación de Serotonina , Animales , Ganglios Basales , Dopamina , Ratones , Serotonina , Sustancia Negra
6.
Cereb Cortex ; 28(6): 1911-1921, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28444137

RESUMEN

Serotonergic neurotransmission, potentially through effects on the brain's default mode network (DMN), may regulate aspects of attention including impulse control. Indeed, genetic variants of the serotonin transporter (5-HTT) have been implicated in impulsivity and related psychopathology. Yet it remains unclear the mechanism by which the 5-HTT genetic variants contribute to individual variability in impulse control. Here, we tested whether DMN connectivity mediates an association between the 5-HTT genetic variants and impulsivity. Participants (N = 92) were from a family cohort study of depression in which we have previously shown a broad distribution of 5-HTT variants. We genotyped for 5-HTTLPR and rs25531 (stratified by transcriptional efficiency: 8 low/low, 53 low/high, and 31 high/high), estimated DMN structural connectivity using diffusion probabilistic tractography, and assessed behavioral measures of impulsivity (from 12 low/low, 48 low/high, and 31 high/high) using the Continuous Performance Task. We found that low transcriptional efficiency genotypes were associated with decreased connection strength between the posterior DMN and the superior frontal gyrus (SFG). Path modeling demonstrated that decreased DMN-SFG connectivity mediated the association between low-efficiency genotypes and increased impulsivity. Taken together, this study suggests a gene-brain-behavior pathway that perhaps underlies the role of the serotonergic neuromodulation in impulse control.


Asunto(s)
Encéfalo/fisiología , Conducta Impulsiva/fisiología , Vías Nerviosas/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Adolescente , Adulto , Anciano , Niño , Estudios de Cohortes , Depresión/genética , Imagen de Difusión Tensora , Femenino , Variación Genética , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
7.
J Neurochem ; 136(3): 526-35, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26484945

RESUMEN

Serotonin receptors are targets of drug therapies for a variety of neuropsychiatric and neurodegenerative disorders. Cocaine inhibits the re-uptake of serotonin (5-HT), dopamine, and noradrenaline, whereas caffeine blocks adenosine receptors and opens ryanodine receptors in the endoplasmic reticulum. We studied how 5-HT and adenosine affected spontaneous GABAergic transmission from thalamic reticular nucleus. We combined whole-cell patch clamp recordings of miniature inhibitory post-synaptic currents (mIPSCs) in ventrobasal thalamic neurons during local (puff) application of 5-HT in wild type (WT) or knockout mice lacking 5-HT2A receptors (5-HT2A -/-). Inhibition of mIPSCs frequency by low (10 µM) and high (100 µM) 5-HT concentrations was observed in ventrobasal neurons from 5-HT2A -/- mice. In WT mice, only 100 µM 5-HT significantly reduced mIPSCs frequency. In 5-HT2A -/- mice, NAN-190, a specific 5-HT1A antagonist, prevented the 100 µM 5-HT inhibition while blocking H-currents that prolonged inhibition during post-puff periods. The inhibitory effects of 100 µM 5-HT were enhanced in cocaine binge-treated 5-HT2A -/- mice. Caffeine binge treatment did not affect 5-HT-mediated inhibition. Our findings suggest that both 5-HT1A and 5-HT2A receptors are present in pre-synaptic thalamic reticular nucleus terminals. Serotonergic-mediated inhibition of GABA release could underlie aberrant thalamocortical physiology described after repetitive consumption of cocaine. Our findings suggest that both 5-HT1A , 5-HT2A and A1 receptors are present in pre-synaptic TRN terminals. 5-HT1A and A1 receptors would down-regulate adenylate cyclase, whereas 5-HT1A would also increase the probability of the opening of G-protein-activated inwardly rectifying K(+) channels (GIRK). Sustained opening of GIRK channels would hyperpolarize pre-synaptic terminals activating H-currents, resulting in less GABA release. 5-HT2A -would activate PLC and IP3 , increasing intracellular [Ca(2+) ] and thus facilitating GABA release.


Asunto(s)
Cafeína/farmacología , Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Receptor de Serotonina 5-HT2A/metabolismo , Núcleos Talámicos/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Cloruro de Cadmio/farmacología , Relación Dosis-Respuesta a Droga , Técnicas In Vitro , Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp , Receptor de Serotonina 5-HT2A/genética , Serotonina/farmacología , Serotoninérgicos/farmacología , Núcleos Talámicos/metabolismo , Fosfolipasas de Tipo C/metabolismo
8.
J Pharmacol Exp Ther ; 357(1): 1-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26787771

RESUMEN

In acute organ injuries, mitochondria are often dysfunctional, and recent research has revealed that recovery of mitochondrial and renal functions is accelerated by induction of mitochondrial biogenesis (MB). We previously reported that the nonselective 5-HT2 receptor agonist DOI [1-(4-iodo-2,5-dimethoxyphenyl)propan-2-amine] induced MB in renal proximal tubular cells (RPTCs). The goal of this study was to determine the role of 5-HT2 receptors in the regulation of mitochondrial genes and oxidative metabolism in the kidney. The 5-HT2C receptor agonist CP-809,101 [2-[(3-chlorophenyl)methoxy]-6-(1-piperazinyl)pyrazine] and antagonist SB-242,084 [6-chloro-2,3-dihydro-5-methyl-N-[6-[(2-methyl-3-pyridinyl)oxy]-3-pyridinyl]-1H-indole-1-carboxyamide dihydrochloride] were used to examine the induction of renal mitochondrial genes and oxidative metabolism in RPTCs and in mouse kidneys in the presence and absence of the 5-HT2C receptor. Unexpectedly, both CP-809,101 and SB-242,084 increased RPTC respiration and peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) mRNA expression in RPTCs at 1-10 nM. In addition, CP-809,101 and SB-242,084 increased mRNA expression of PGC-1α and the mitochondrial proteins NADH dehydrogenase subunit 1 and NADH dehydrogenase (ubiquinone) ß subcomplex 8 in mice. These compounds increased mitochondrial genes in RPTCs in which the 5-HT2C receptor was downregulated with small interfering RNA and in the renal cortex of mice lacking the 5-HT2C receptor. By contrast, the ability of these compounds to increase PGC-1α mRNA and respiration was blocked in RPTCs treated with 5-HT2A receptor small interfering RNA or the 5-HT2A receptor antagonist eplivanserin. In addition, the 5-HT2A receptor agonist NBOH-2C-CN [4-[2-[[(2-hydroxyphenyl)methyl]amino]ethyl]-2,5-dimethoxybenzonitrile] increased RPTC respiration at 1-100 nM. These results suggest that agonism of the 5-HT2A receptor induces MB and that the classic 5-HT2C receptor agonist CP-809,101 and antagonist SB-242,084 increase mitochondrial genes and oxidative metabolism through the 5-HT2A receptor. To our knowledge, this is the first report that links 5-HT2A receptor agonism to mitochondrial function.


Asunto(s)
Mitocondrias/genética , Receptor de Serotonina 5-HT2A/efectos de los fármacos , Receptor de Serotonina 5-HT2A/genética , Agonistas del Receptor de Serotonina 5-HT2/farmacología , Antagonistas del Receptor de Serotonina 5-HT2/farmacología , Aminopiridinas/farmacología , Animales , Complejo I de Transporte de Electrón/biosíntesis , Complejo I de Transporte de Electrón/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Indoles/farmacología , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Oxidación-Reducción , Consumo de Oxígeno , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Piperazinas/farmacología , Pirazinas/farmacología , Conejos , Receptor de Serotonina 5-HT2C/efectos de los fármacos , Receptor de Serotonina 5-HT2C/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
9.
J Neurosci ; 34(17): 6107-11, 2014 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-24760870

RESUMEN

The activity of the prefrontal cortex is essential for normal emotional processing and is strongly modulated by serotonin (5-HT). Yet, little is known about the regulatory mechanisms that control the activity of the prefrontal 5-HT receptors. Here, we found and characterized a deregulation of prefrontal 5-HT receptor electrophysiological signaling in mouse models of disrupted serotonin transporter (5-HTT) function, a risk factor for emotional and cognitive disturbances. We identified a novel tyrosine kinase-dependent mechanism that regulates 5-HT-mediated inhibition of prefrontal pyramidal neurons. We report that mice with compromised 5-HTT, resulting from either genetic deletion or brief treatment with selective serotonin reuptake inhibitors during development, have amplified 5-HT1A receptor-mediated currents in adulthood. These greater inhibitory effects of 5-HT are accompanied by enhanced downstream coupling to Kir3 channels. Notably, in normal wild-type mice, we found that these larger 5-HT1A responses can be mimicked through inhibition of Src family tyrosine kinases. By comparison, in our 5-HTT mouse models, the larger 5-HT1A responses were rapidly reduced through inhibition of tyrosine phosphatases. Our findings implicate tyrosine phosphorylation in regulating the electrophysiological effects of prefrontal 5-HT1A receptors with implications for neuropsychiatric diseases associated with emotional dysfunction, such as anxiety and depressive disorders.


Asunto(s)
Conducta Animal/fisiología , Corteza Prefrontal/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Serotonina/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Femenino , Inhibición Psicológica , Masculino , Ratones , Fosforilación/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Transmisión Sináptica/efectos de los fármacos
10.
J Neurosci ; 34(37): 12379-93, 2014 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-25209278

RESUMEN

Early-life serotonin [5-hydroxytryptamine (5-HT)] signaling modulates brain development, which impacts adult behavior, but 5-HT-sensitive periods, neural substrates, and behavioral consequences remain poorly understood. Here we identify the period ranging from postnatal day 2 (P2) to P11 as 5-HT sensitive, with 5-HT transporter (5-HTT) blockade increasing anxiety- and depression-like behavior, and impairing fear extinction learning and memory in adult mice. Concomitantly, P2-P11 5-HTT blockade causes dendritic hypotrophy and reduced excitability of infralimbic (IL) cortex pyramidal neurons that normally promote fear extinction. By contrast, the neighboring prelimbic (PL) pyramidal neurons, which normally inhibit fear extinction, become more excitable. Excitotoxic IL but not PL lesions in adult control mice reproduce the anxiety-related phenotypes. These findings suggest that increased 5-HT signaling during P2-P11 alters adult mPFC function to increase anxiety and impair fear extinction, and imply a differential role for IL and PL neurons in regulating affective behaviors. Together, our results support a developmental mechanism for the etiology and pathophysiology of affective disorders and fear-related behaviors.


Asunto(s)
Envejecimiento/metabolismo , Ansiedad/metabolismo , Depresión/metabolismo , Extinción Psicológica , Miedo , Corteza Prefrontal/fisiopatología , Serotonina/metabolismo , Animales , Animales Recién Nacidos , Ansiedad/complicaciones , Conducta Animal , Depresión/complicaciones , Femenino , Masculino , Ratones
11.
Nature ; 452(7183): 93-7, 2008 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-18297054

RESUMEN

The psychosis associated with schizophrenia is characterized by alterations in sensory processing and perception. Some antipsychotic drugs were identified by their high affinity for serotonin 5-HT2A receptors (2AR). Drugs that interact with metabotropic glutamate receptors (mGluR) also have potential for the treatment of schizophrenia. The effects of hallucinogenic drugs, such as psilocybin and lysergic acid diethylamide, require the 2AR and resemble some of the core symptoms of schizophrenia. Here we show that the mGluR2 interacts through specific transmembrane helix domains with the 2AR, a member of an unrelated G-protein-coupled receptor family, to form functional complexes in brain cortex. The 2AR-mGluR2 complex triggers unique cellular responses when targeted by hallucinogenic drugs, and activation of mGluR2 abolishes hallucinogen-specific signalling and behavioural responses. In post-mortem human brain from untreated schizophrenic subjects, the 2AR is upregulated and the mGluR2 is downregulated, a pattern that could predispose to psychosis. These regulatory changes indicate that the 2AR-mGluR2 complex may be involved in the altered cortical processes of schizophrenia, and this complex is therefore a promising new target for the treatment of psychosis.


Asunto(s)
Trastornos Psicóticos/metabolismo , Receptor de Serotonina 5-HT2A/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Línea Celular , Células Cultivadas , Regulación hacia Abajo , Alucinógenos/metabolismo , Alucinógenos/farmacología , Humanos , Ratones , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Trastornos Psicóticos/tratamiento farmacológico , Trastornos Psicóticos/genética , Receptor de Serotonina 5-HT2A/análisis , Receptor de Serotonina 5-HT2A/deficiencia , Receptor de Serotonina 5-HT2A/genética , Receptores de Glutamato Metabotrópico/análisis , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Receptores de Glutamato Metabotrópico/genética , Esquizofrenia/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba
12.
Genome Res ; 20(7): 972-80, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20488932

RESUMEN

Abnormalities of genomic methylation patterns are lethal or cause disease, but the cues that normally designate CpG dinucleotides for methylation are poorly understood. We have developed a new method of methylation profiling that has single-CpG resolution and can address the methylation status of repeated sequences. We have used this method to determine the methylation status of >275 million CpG sites in human and mouse DNA from breast and brain tissues. Methylation density at most sequences was found to increase linearly with CpG density and to fall sharply at very high CpG densities, but transposons remained densely methylated even at higher CpG densities. The presence of histone H2A.Z and histone H3 di- or trimethylated at lysine 4 correlated strongly with unmethylated DNA and occurred primarily at promoter regions. We conclude that methylation is the default state of most CpG dinucleotides in the mammalian genome and that a combination of local dinucleotide frequencies, the interaction of repeated sequences, and the presence or absence of histone variants or modifications shields a population of CpG sites (most of which are in and around promoters) from DNA methyltransferases that lack intrinsic sequence specificity.


Asunto(s)
Secuencia de Bases/fisiología , Cromatina/química , Cromatina/fisiología , Metilación de ADN , Animales , Encéfalo/metabolismo , Mama/metabolismo , Cromatina/genética , Mapeo Cromosómico , Islas de CpG/genética , Femenino , Genoma , Histonas/metabolismo , Humanos , Ratones , Análisis de Secuencia de ADN , Estudios de Validación como Asunto
13.
Child Adolesc Psychiatry Ment Health ; 17(1): 73, 2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37328889

RESUMEN

BACKGROUND: Selective serotonin reuptake inhibitors (SSRIs) are the most frequently prescribed antidepressants in pregnancy. Animal and some clinical studies have suggested potential increases in depression and anxiety following prenatal SSRI exposure, but the extent to which these are driven by the medication remains unclear. We used Danish population data to test associations between maternal SSRI use during pregnancy and children outcomes up to age 22. METHODS: We prospectively followed 1,094,202 single-birth Danish children born 1997-2015. The primary exposure was ≥ 1 SSRI prescription filled during pregnancy; the primary outcome, first diagnosis of a depressive, anxiety, or adjustment disorder, or redeemed prescription for an antidepressant medication. We used propensity score weights to adjust potential confounders, and incorporated data from the Danish National Birth Cohort (1997-2003) to further quantify potential residual confounding by subclinical factors. RESULTS: The final dataset included 15,651 exposed and 896,818 unexposed, children. After adjustments, SSRI-exposed had higher rates of the primary outcome than those of mothers who either did not use an SSRI (HR = 1.55 [95%CI:1.44,1.67] or discontinued the SSRI use ≥ 3 months prior to conception (HR = 1.23 [1.13,1.34]). Age of onset was earlier among exposed (9 [IQR:7-13] years) versus unexposed (12 [IQR:12-17] years) children (p < 0.01). Paternal SSRI use in the absence of maternal use during the index pregnancy (HR = 1.46 [1.35,1.58]) and maternal SSRI use only after pregnancy (HR = 1.42 [1.35,1.49]) were each also associated with these outcomes. CONCLUSIONS: While SSRI exposure was associated with increased risk in the children, this risk may be driven at least partly by underlying severity of maternal illness or other confounding factors.

14.
J Dev Orig Health Dis ; 14(5): 591-601, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37732425

RESUMEN

The deleterious effects of adversity are likely intergenerational, such that one generation's adverse experiences can affect the next. Epidemiological studies link maternal adversity to offspring depression and anxiety, possibly via transmission mechanisms that influence offspring fronto-limbic connectivity. However, studies have not thoroughly disassociated postnatal exposure effects nor considered the role of offspring sex. We utilized infant neuroimaging to test the hypothesis that maternal childhood maltreatment (CM) would be associated with increased fronto-limbic connectivity in infancy and tested brain-behavior associations in childhood. Ninety-two dyads participated (32 mothers with CM, 60 without; 52 infant females, 40 infant males). Women reported on their experiences of CM and non-sedated sleeping infants underwent MRIs at 2.44 ± 2.74 weeks. Brain volumes were estimated via structural MRI and white matter structural connectivity (fiber counts) via diffusion MRI with probabilistic tractography. A subset of parents (n = 36) reported on children's behaviors at age 5.17 ± 1.73 years. Males in the maltreatment group demonstrated greater intra-hemispheric fronto-limbic connectivity (b = 0.96, p= 0.008, [95%CI 0.25, 1.66]), no differences emerged for females. Fronto-limbic connectivity was related to somatic complaints in childhood only for males (r = 0.673, p = 0.006). Our findings suggest that CM could have intergenerational associations to offspring brain development, yet mechanistic studies are needed.


Asunto(s)
Sustancia Blanca , Masculino , Lactante , Niño , Humanos , Femenino , Preescolar , Sustancia Blanca/diagnóstico por imagen , Madres , Encéfalo/diagnóstico por imagen , Imagen por Resonancia Magnética , Ansiedad
15.
J Neurosci ; 31(44): 15742-50, 2011 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-22049417

RESUMEN

Serotonin (5-HT)-selective reuptake inhibitors (SSRIs) are widely administered for the treatment of depression, anxiety, and other neuropsychiatric disorders, but response rates are low, and side effects often lead to discontinuation. Side effect profiles suggest that SSRIs inhibit dopaminergic activity, but mechanistic insight remains scarce. Here we show that in mice, chronic 5-HT transporter (5-HTT) blockade during adulthood but not during development impairs basal ganglia-dependent behaviors in a dose-dependent and reversible fashion. Furthermore, chronic 5-HTT blockade reduces striatal dopamine (DA) content and metabolism. A causal relationship between reduced DA signaling and impaired basal ganglia-dependent behavior is indicated by the reversal of behavioral deficits through L-DOPA administration. Our data suggest that augmentation of DA signaling would reduce side effects and increase efficacies of SSRI-based therapy.


Asunto(s)
Ganglios Basales/efectos de los fármacos , Dopamina/metabolismo , Fluoxetina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Transducción de Señal/efectos de los fármacos , Ácido 3,4-Dihidroxifenilacético/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Ganglios Basales/patología , Ganglios Basales/fisiopatología , Recuento de Células , Cromatografía Líquida de Alta Presión/métodos , Dopaminérgicos/farmacología , Dopaminérgicos/uso terapéutico , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Interacciones Farmacológicas , Conducta Exploratoria/efectos de los fármacos , Fluoxetina/uso terapéutico , Ácido Homovanílico/metabolismo , Levodopa/farmacología , Levodopa/uso terapéutico , Masculino , Ratones , Ratones Transgénicos , Trastornos del Movimiento/tratamiento farmacológico , Trastornos del Movimiento/genética , Trastornos del Movimiento/patología , Desempeño Psicomotor/efectos de los fármacos , Distribución Aleatoria , Prueba de Desempeño de Rotación con Aceleración Constante , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Transducción de Señal/fisiología , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Sustancia Negra/patología , Área Tegmental Ventral/citología , Área Tegmental Ventral/efectos de los fármacos
16.
Neuron ; 53(3): 439-52, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17270739

RESUMEN

Hallucinogens, including mescaline, psilocybin, and lysergic acid diethylamide (LSD), profoundly affect perception, cognition, and mood. All known drugs of this class are 5-HT(2A) receptor (2AR) agonists, yet closely related 2AR agonists such as lisuride lack comparable psychoactive properties. Why only certain 2AR agonists are hallucinogens and which neural circuits mediate their effects are poorly understood. By genetically expressing 2AR only in cortex, we show that 2AR-regulated pathways on cortical neurons are sufficient to mediate the signaling pattern and behavioral response to hallucinogens. Hallucinogenic and nonhallucinogenic 2AR agonists both regulate signaling in the same 2AR-expressing cortical neurons. However, the signaling and behavioral responses to the hallucinogens are distinct. While lisuride and LSD both act at 2AR expressed by cortex neurons to regulate phospholipase C, LSD responses also involve pertussis toxin-sensitive heterotrimeric G(i/o) proteins and Src. These studies identify the long-elusive neural and signaling mechanisms responsible for the unique effects of hallucinogens.


Asunto(s)
Conducta Animal/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Alucinógenos/farmacología , Receptor de Serotonina 5-HT2A/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Anfetaminas , Animales , Autorradiografía , Unión Competitiva/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Electrofisiología , Hibridación Fluorescente in Situ , Ketanserina/farmacología , Lisurida/farmacología , Masculino , Ratones , Ratones Noqueados , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Receptor de Serotonina 5-HT2A/genética , Receptores de Dopamina D1/efectos de los fármacos , Receptores de Dopamina D2/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Antagonistas de la Serotonina/farmacología , Agonistas de Receptores de Serotonina/farmacología
17.
Sci Rep ; 11(1): 22852, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34819526

RESUMEN

Depression and anxiety, two of the most common mental health disorders, share common symptoms and treatments. Most pharmacological agents available to treat these disorders target monoamine systems. Currently, finding the most effective treatment for an individual is a process of trial and error. To better understand how disease etiology may predict treatment response, we studied mice exposed developmentally to the selective serotonin reuptake inhibitor (SSRI) fluoxetine (FLX). These mice show the murine equivalent of anxiety- and depression-like symptoms in adulthood and here we report that these mice are also behaviorally resistant to the antidepressant-like effects of adult SSRI administration. We investigated whether tianeptine (TIA), which exerts its therapeutic effects through agonism of the mu-opioid receptor instead of targeting monoaminergic systems, would be more effective in this model. We found that C57BL/6J pups exposed to FLX from postnatal day 2 to 11 (PNFLX, the mouse equivalent in terms of brain development to the human third trimester) showed increased avoidant behaviors as adults that failed to improve, or were even exacerbated, by chronic SSRI treatment. By contrast, avoidant behaviors in these same mice were drastically improved following chronic treatment with TIA. Overall, this demonstrates that TIA may be a promising alternative treatment for patients that fail to respond to typical antidepressants, especially in patients whose serotonergic system has been altered by in utero exposure to SSRIs.


Asunto(s)
Antidepresivos de Segunda Generación/toxicidad , Antidepresivos Tricíclicos/farmacología , Reacción de Prevención/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Fluoxetina/toxicidad , Inhibidores Selectivos de la Recaptación de Serotonina/toxicidad , Tiazepinas/farmacología , Animales , Animales Recién Nacidos , Encéfalo/crecimiento & desarrollo , Conducta Alimentaria/efectos de los fármacos , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Prueba de Campo Abierto/efectos de los fármacos
18.
Artículo en Inglés | MEDLINE | ID: mdl-33487578

RESUMEN

BACKGROUND: Major depressive disorder (MDD) is associated with aberrant limbic neural responses to emotional stimuli. We assessed how self-generated emotions modulate trial-by-trial limbic activity and whether this brain-emotion synchrony varies by familial MDD risk (regardless of personal MDD history) and neuroticism. METHODS: Participants (n = 74, mean age = 34 years) were later-generation family members of depressed or nondepressed probands as part of a longitudinal cohort study. Using an emotion induction task, we examined participant-specific modulation of anatomically defined limbic neurobiology. Neuroticism, mental health, and familial parenting style were assessed, and MDD assessments were routinely collected throughout the previous longitudinal assessments of the study. RESULTS: Participant-specific emotional arousal modulated amygdala and hippocampal activity. Lasso regression identified attenuated right amygdala arousal modulation as being relatively more associated with neuroticism (even though neuroticism was not associated with arousal ratings). Attenuated amygdala modulation and neuroticism were significantly more likely in offspring of parents with MDD. Parental MDD, but not personal history of MDD, predicted attenuated amygdala modulation. CONCLUSIONS: Attenuated right amygdala modulation by emotional arousal was associated with neuroticism, indicating that the amygdala was less synchronous with emotional experiences in individuals higher in neuroticism. This neurophenotype was predicted by participants' parental MDD history but not by their own MDD history; that is, it was observed in unaffected and affected offspring of parents with MDD. These data suggest that weak amygdala-emotion synchrony may be a predisposing risk factor for MDD, rather than a result of the illness, and they suggest pathways by which this risk factor for depression is passed intergenerationally.


Asunto(s)
Trastorno Depresivo Mayor , Adulto , Amígdala del Cerebelo , Depresión , Emociones , Humanos , Estudios Longitudinales , Imagen por Resonancia Magnética
19.
Int J Neuropsychopharmacol ; 13(10): 1299-314, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20158933

RESUMEN

Atypical antipsychotic drugs (APDs) increase dopamine (DA) release in prefrontal cortex (PFC), an effect probably mediated by the direct or indirect activation of the 5-HT(1A) receptor (5-HT(1A)R). Given the very low in-vitro affinity of most APDs for 5-HT(1A)Rs and the large co-expression of 5-HT(1A)Rs and 5-HT(2A) receptors (5-HT(2A)Rs) in the PFC, this effect might result from the imbalance of 5-HT(1A)R and 5-HT(2A)R activation after blockade of these receptors by APDs, for which they show high affinity. Here we tested this hypothesis by examining the dependence of the APD-induced DA release in medial PFC (mPFC) on each receptor by using in-vivo microdialysis in wild-type (WT) and 5-HT(1A)R and 5-HT(2A)R knockout (KO) mice. Local APDs (clozapine, olanzapine, risperidone) administered by reverse dialysis induced a dose-dependent increase in mPFC DA output equally in WT and 5-HT(2A)R KO mice whereas the DA increase was absent in 5-HT(1A)R KO mice. To examine the relative contribution of both receptors to the clozapine-induced DA release in rat mPFC, we silenced G-protein-coupled receptors (GPCRs) in vivo with N-ethoxycarbonyl-2-ethoxy-1,2-dihydroquinoline (EEDQ) while 5-HT(1A)Rs or 5-HT(2A)/2CRs in the mPFC were selectively protected with the respective antagonists WAY-100635 or ritanserin. The inactivation of GPCRs while preserving ∼70% of 5-HT(2A)/(2C)Rs prevented the clozapine-induced DA rise in mPFC. In contrast, clozapine increased DA in mPFC of EEDQ-treated rats whose 5-HT(1A)Rs were protected (∼50% of control rats). These results indicate that (1) 5-HT(1A)Rs are necessary for the APDs-induced elevation in cortical DA transmission, and (2) this effect does not require 5-HT(2A)R blockade by APDs.


Asunto(s)
Antipsicóticos/farmacología , Dopamina/metabolismo , Corteza Prefrontal/efectos de los fármacos , Receptor de Serotonina 5-HT1A/metabolismo , Receptor de Serotonina 5-HT2A/metabolismo , Animales , Benzodiazepinas/farmacología , Clozapina/farmacología , Antagonistas de Dopamina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microdiálisis , Modelos Animales , Olanzapina , Quinolinas/metabolismo , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Risperidona/farmacología , Esquizofrenia/tratamiento farmacológico
20.
J Neurosci ; 28(1): 199-207, 2008 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-18171937

RESUMEN

Serotonin (5-HT) acts as a neurotransmitter, but also modulates brain maturation during early development. The demonstrated influence of genetic variants on brain function, personality traits, and susceptibility to neuropsychiatric disorders suggests a critical importance of developmental mechanisms. However, little is known about how and when developmentally perturbed 5-HT signaling affects circuitry and resulting behavior. The 5-HT transporter (5-HTT) is a key regulator of extracellular 5-HT levels and we used pharmacologic strategies to manipulate 5-HTT function during development and determine behavioral consequences. Transient exposure to the 5-HTT inhibitors fluoxetine, clomipramine, and citalopram from postnatal day 4 (P4) to P21 produced abnormal emotional behaviors in adult mice. Similar treatment with the norepinephrine transporter (NET) inhibitor, desipramine, did not adversely affect adult behavior, suggesting that 5-HT and norepinephrine (NE) do not share the same effects on brain development. Shifting our period of treatment/testing to P90/P185 failed to mimic the effect of earlier exposure, demonstrating that 5-HT effects on adult behavior are developmentally specific. We have hypothesized that early-life perturbations of 5-HT signaling affect corticolimbic circuits that do not reach maturity until the peri-adolescent period. In support of this idea, we found that abnormal behaviors resulting from postnatal fluoxetine exposure have a post-pubescent onset and persist long after reaching adult age. A better understanding of the underlying 5-HT sensitive circuits and how they are perturbed should lead to new insights into how various genetic polymorphisms confer their risk to carriers. Furthermore, these studies should help determine whether in utero exposure to 5-HTT blocking drugs poses a risk for behavioral abnormalities in later life.


Asunto(s)
Emociones , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/fisiología , Norepinefrina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Serotonina/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Antidepresivos/metabolismo , Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Emociones/efectos de los fármacos , Emociones/fisiología , Reacción de Fuga/efectos de los fármacos , Reacción de Fuga/fisiología , Conducta Exploratoria/fisiología , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/fisiología , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Noqueados , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA